Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems
Abstract
:1. Introduction
Cancer Statistics: Need for Better Therapeutics
2. Limitations and Challenges Associated with Traditional Anticancer Therapies
2.1. Cancer Surgery
2.2. Chemotherapy
2.3. Radiotherapy
3. Targeted Drug Delivery Systems and Their Limitations
3.1. Active Tumor Targeting Approach
3.1.1. Antibody and Antibody Fragments
3.1.2. Immunotoxins and Immunoconjugates
3.1.3. Immunoliposomes
3.1.4. Manufacturing and Clinical Challenges of Active Targeting
3.2. Passive Tumor Targeting Approach
3.2.1. Traditional Liposomes
3.2.2. Stealth Liposome
3.2.3. Requirements of Stimuli Induced Drug Release
Ultrasound Induced Triggered Release
Light Induced Triggered Release
Hyperthermia Induced Triggered Release
Triggered Release by Magnetic Field from Magnetic Liposomes
3.3. Manufacturing and Clinical Challenges Common to Both Active and Passive Targeting
4. Need for Better Pre-Clinical and Clinical Strategies
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Available online: https://web.archive.org/web/20210707200955/https://gicr.iarc.fr/public/docs/20120906-WorldCancerFactSheet.pdf (accessed on 7 July 2021).
- Available online: https://web.archive.org/web/20210707201547/https://gco.iarc.fr/today/data/factsheets/cancers/39-All-cancers-fact-sheet.pdf (accessed on 7 July 2021).
- Available online: https://web.archive.org/web/20210707201915/https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2021/cancer-facts-and-figures-2021.pdf (accessed on 7 July 2021).
- Gyanani, V. Turning Stealth Liposomes into Cationic Liposomes for Anticancer Drug Delivery. Ph.D. Thesis, University of the Pacific, Stockton, CA, USA, 2013. [Google Scholar]
- Allen, T.M. Ligand-targeted therapeutics in anticancer therapy. Nat. Rev. Cancer 2002, 2, 750–763. [Google Scholar] [CrossRef]
- Yao, X.; Panichpisal, K.; Kurtzman, N.; Nugent, K. Cisplatin nephrotoxicity: A review. Am. J. Med. Sci. 2007, 334, 115–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pfeffer, B.; Tziros, C.; Katz, R.J. Current concepts of anthracycline cardiotoxicity: Pathogenesis, diagnosis and prevention. Br. J. Cardiol. 2009, 16, 85–89. [Google Scholar]
- Partridge, A.H.; Burstein, H.J.; Winer, E.P. Side effects of chemotherapy and combined chemohormonal therapy in women with early-stage breast cancer. J. Natl. Cancer Inst. Monogr. 2001, 30, 135–142. [Google Scholar] [CrossRef] [PubMed]
- Fabbrocini, G.; Cameli, N.; Romano, M.C.; Mariano, M.; Panariello, L.; Bianca, D.; Giuseppe, M. Chemotherapy and skin reactions. J. Exp. Clin. Cancer Res. 2012, 31, 50. [Google Scholar] [CrossRef] [Green Version]
- Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug resistance in cancer: Role of ATP–dependent transporters. Nat. Rev. Cancer 2002, 2, 48–58. [Google Scholar] [CrossRef] [Green Version]
- Bentzen, S.M. Preventing or reducing late side effects of radiation therapy: Radiobiology meets molecular pathology. Nat. Rev. Cancer 2006, 6, 702–713. [Google Scholar] [CrossRef]
- Bosslet, K.; Straub, R.; Blumrich, M.; Czech, J.; Gerken, M.; Sperker, B.; Kroemer, H.K.; Gesson, J.P.; Koch, M.; Monneret, C. Elucidation of the mechanism enabling tumor selective prodrug monotherapy. Cancer Res. 1998, 58, 1195–1201. [Google Scholar]
- Senzer, N.; Nemunaitis, J.; Nemunaitis, D.; Bedell, C.; Edelman, G.; Barve, M.; Nunan, R.; Pirollo, K.F.; Rait, A.; Chang, E.H. Results of a Phase I Trial of SGT-53: A Systemically Administered, Tumor-Targeting Immunoliposome Nanocomplex Incorporating a Plasmid Encoding wtp53. Clin. Gene Cell Ther. Oral Abstr. Sess. 2012, 20. [Google Scholar] [CrossRef]
- Pirollo, K.F.; Nemunaitis, J.; Leung, P.K.; Nunan, R.; Adams, J.; Chang, E.H. Safety and Efficacy in Advanced Solid Tumors of a Targeted Nanocomplex Carrying the p53 Gene Used in Combination with Docetaxel: A Phase 1b Study. Mol. Ther. 2016, 24, 1697–1706. [Google Scholar] [CrossRef] [Green Version]
- Kim, E.M.; Jeong, H.J. Liposomes: Biomedical Applications. Chonnam. Med. J. 2021, 57, 27–35. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://clinicaltrials.gov/ct2/show/NCT03076372 (accessed on 12 August 2021).
- Moles, E.; Kavallaris, M. A potent targeted cancer nanotherapeutic. Nat. Biomed. Eng. 2019, 3, 248–250. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; Sun, Y.; Liu, Y.; Meng, F.; Lee, R.J. Clinical translation of immunoliposomes for cancer therapy: Recent perspectives. Expert Opin. Drug Deliv. 2018, 15, 893–903. [Google Scholar] [CrossRef]
- Yan, W.; Leung, S.S.; To, K.K. Updates on the use of liposomes for active tumor targeting in cancer therapy. Nanomedicine 2020, 15, 303–318. [Google Scholar] [CrossRef]
- Available online: https://clinicaltrials.gov/ct2/show/NCT03603379 (accessed on 12 August 2021).
- Paranthaman, S.; Goravinahalli Shivananjegowda, M.; Mahadev, M.; Moin, A.; Hagalavadi Nanjappa, S.; Nanjaiyah, N.; Chidambaram, S.B.; Gowda, D.V. Nanodelivery Systems Targeting Epidermal Growth Factor Receptors for Glioma Management. Pharmaceutics 2020, 12, 1198. [Google Scholar] [CrossRef]
- Miller, K.; Cortes, J.; Hurvitz, S.A.; Krop, I.E.; Tripathy, D.; Verma, S.; Riahi, K.; Reynolds, J.G.; Wickham, T.J.; Molnar, I.; et al. HERMIONE: A randomized Phase 2 trial of MM-302 plus trastuzumab versus chemotherapy of physician’s choice plus trastuzumab in patients with previously treated, anthracycline-naïve, HER2-positive, locally advanced/metastatic breast cancer. BMC Cancer 2016, 16, 352. [Google Scholar] [CrossRef] [Green Version]
- Available online: https://www.clinicaltrials.gov/ct2/show/NCT01304797 (accessed on 12 August 2021).
- Gargett, T.; Abbas, M.N.; Rolan, P.; Price, J.D.; Gosling, K.M.; Ferrante, A.; Ruszkiewicz, A.; Atmosukarto, I.I.C.; Altin, J.; Parish, C.R.; et al. Phase I trial of Lipovaxin-MM, a novel dendritic cell-targeted liposomal vaccine for malignant melanoma. Cancer Immunol. Immunother. 2018, 67, 1461–1472. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Zhang, L.; Zhu, W.; Guo, R.; Sun, H.; Chen, X.; Deng, N. Barriers and Strategies of Cationic Liposomes for Cancer Gene Therapy. Mol. Ther.-Methods Clin. Dev. 2020, 18, 751–764. [Google Scholar] [CrossRef]
- Available online: https://www.clinicaltrials.gov/ct2/show/NCT01517464 (accessed on 12 August 2021).
- Siefker-Radtke, A.; Zhang, X.Q.; Guo, C.C.; Shen, Y.; Pirollo, K.F.; Sabir, S.; Leung, C.; Leong-Wu, C.; Ling, C.M.; Chang, E.H.; et al. A Phase l Study of a Tumor-targeted Systemic Nanodelivery System, SGT-94, in Genitourinary Cancers. Mol. Ther. 2016, 24, 1484–1491. [Google Scholar] [CrossRef] [Green Version]
- Available online: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/748730 (accessed on 12 August 2021).
- Available online: https://patents.justia.com/patent/20180334724 (accessed on 12 August 2021).
- Song, X.L.; Liu, S.; Jiang, Y.; Gu, L.Y.; Xiao, Y.; Wang, X.; Cheng, L.; Li, X.T. Targeting vincristine plus tetrandrine liposomes modified with DSPE-PEG(2000)-transferrin in treatment of brain glioma. Eur. J. Pharm. Sci. 2017, 96, 129–140. [Google Scholar] [CrossRef]
- Chiani, M.; Norouzian, D.; Shokrgozar, M.A.; Azadmanesh, K.; Najmafshar, A.; Mehrabi, M.R.; Akbarzadeh, A. Folic acid conjugated nanoliposomes as promising carriers for targeted delivery of bleomycin. Artif. Cells Nanomed. Biotechnol. 2018, 46, 757–763. [Google Scholar] [CrossRef] [PubMed]
- Della Giovampaola, C.; Capone, A.; Ermini, L.; Lupetti, P.; Vannuccini, E.; Finetti, F.; Donnini, S.; Ziche, M.; Magnani, A.; Leone, G.; et al. Formulation of liposomes functionalized with Lotus lectin and effective in targeting highly proliferative cells. Biochim Biophys. Acta Gen. Subj. 2017, 1861, 860–870. [Google Scholar] [CrossRef] [PubMed]
- Cristiano, M.C.; Cosco, D.; Celia, C.; Tudose, A.; Mare, R.; Paolino, D.; Fresta, M. Anticancer activity of all-trans retinoic acid-loaded liposomes on human thyroid carcinoma cells. Colloids Surf. B Biointerfaces 2017, 150, 408–416. [Google Scholar] [CrossRef]
- Grace, V.M.; Viswanathan, S. Pharmacokinetics and therapeutic efficiency of a novel cationic liposome nano-formulated all trans retinoic acid in lung cancer mice model. J. Drug Deliv. Sci. Technol. 2017, 39, 223–236. [Google Scholar] [CrossRef]
- Lancet, J.E.; Uy, G.L.; Cortes, J.E.; Newell, L.F.; Lin, T.L.; Ritchie, E.K.; Stuart, R.K.; Strickland, S.A.; Hogge, D.; Solomon, S.R.; et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients with Newly Diagnosed Secondary Acute Myeloid Leukemia. J. Clin. Oncol. 2018, 36, 2684–2692. [Google Scholar] [CrossRef] [PubMed]
- Pelzer, U.; Blanc, J.F.; Melisi, D.; Cubillo, A.; Von Hoff, D.D.; Wang-Gillam, A.; Chen, L.T.; Siveke, J.T.; Wan, Y.; Solem, C.T.; et al. Quality-adjusted survival with combination nal-IRI+5-FU/LV vs. 5-FU/LV alone in metastatic pancreatic cancer patients previously treated with gemcitabine-based therapy: A Q-TWiST analysis. Br. J. Cancer 2017, 116, 1247–1253. [Google Scholar] [CrossRef] [Green Version]
- Tran, S.; DeGiovanni, P.J.; Piel, B.; Rai, P. Cancer nanomedicine: A review of recent success in drug delivery. Clin. Transl. Med. 2017, 6, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal Formulations in Clinical Use: An Updated Review. Pharmaceutics 2017, 9, 12. [Google Scholar] [CrossRef]
- Lamichhane, N.; Udayakumar, T.S.; D’Souza, W.D.; Simone, C.B., 2nd; Raghavan, S.R.; Polf, J.; Mahmood, J. Liposomes: Clinical Applications and Potential for Image-Guided Drug Delivery. Molecules 2018, 23, 288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bozzuto, G.; Molinari, A. Liposomes as nanomedical devices. Int. J. Nanomed. 2015, 10, 975–999. [Google Scholar] [CrossRef] [Green Version]
- Available online: http://web.archive.org/web/20210813002334/https://patents.google.com/patent/AU2013347990A1/en (accessed on 13 August 2021).
- Available online: http://web.archive.org/web/20210813001653/http://www.druginformation.com/rxdrugs/V/VinCRIStine%20Sulfate%20LIPOSOME%20Injection.html (accessed on 13 August 2021).
- Available online: http://web.archive.org/web/20210813003036/https://www.bausch.com/ecp/our-products/rx-pharmaceuticals/rx-pharmaceuticals/visudyne-verteporfin-for-injection (accessed on 13 August 2021).
- Ghosh, S.; Carter, K.A.; Lovell, J.F. Liposomal formulations of photosensitizers. Biomaterials 2019, 218, 119341. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://web.archive.org/web/20210422023716/https://clinicaltrials.gov/ct2/show/NCT04590664 (accessed on 13 August 2021).
- Available online: https://web.archive.org/web/20210813005341/https://clinicaltrials.gov/ct2/show/NCT00617981 (accessed on 13 August 2021).
- Lombardo, D.; Calandra, P.; Barreca, D.; Magazù, S.; Kiselev, M.A. Soft Interaction in Liposome Nanocarriers for Therapeutic Drug Delivery. Nanomaterials 2016, 6, 125. [Google Scholar] [CrossRef] [PubMed]
- Available online: http://web.archive.org/web/20210813010747/https://www.syncorebio.com/en/sb05pc-endotag-1-phase-iii-been-approved-in-china-by-nmpa/ (accessed on 13 August 2021).
- Hong, D.S.; Kang, Y.-K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumors. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef] [PubMed]
- Lin, X.; Chen, W.; Wei, F.; Zhou, B.P.; Hung, M.C.; Xie, X. Nanoparticle Delivery of miR-34a Eradicates Long-term-cultured Breast Cancer Stem Cells via Targeting C22ORF28 Directly. Theranostics 2017, 7, 4805–4824. [Google Scholar] [CrossRef] [Green Version]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
- Available online: https://web.archive.org/web/20210813021139/https://www.onivyde.com/websites/onivyde_us_online/wp-content/uploads/sites/3/2018/12/14105740/ONIVYDE_USPI.pdf (accessed on 13 August 2021).
- Available online: http://web.archive.org/web/20210225052514if_/https://www.curetoday.com/view/onivyde-shows-promise-in-patients-with-small-cell-lung-cancer-who-become-resistant-to-chemotherapy (accessed on 13 August 2021).
- Gabizon, A.; Shmeeda, H.; Tahover, E.; Kornev, G.; Patil, Y.; Amitay, Y.; Ohana, P.; Sapir, E.; Zalipsky, S. Development of Promitil®, a lipidic prodrug of mitomycin c in PEGylated liposomes: From bench to bedside. Adv. Drug Deliv. Rev. 2020, 154–155, 13–26. [Google Scholar] [CrossRef]
- Available online: http://lipomedix.com/Products/%C2%AEPromitil (accessed on 12 August 2021).
- Available online: http://web.archive.org/web/20210813024839/https://www.globenewswire.com/news-release/2020/01/23/1974347/0/en/LipoMedix-Announces-Publication-of-Positive-Phase-1-Data-for-Promitil-PL-MLP-in-Research-Journal-Investigational-New-Drugs.html (accessed on 13 August 2021).
- Available online: https://web.archive.org/web/20210813025653/https://www.genprex.com/technology/reqorsa/ (accessed on 13 August 2021).
- Available online: https://web.archive.org/web/20210813030047/https://adisinsight.springer.com/drugs/800018766 (accessed on 13 August 2021).
- Available online: https://web.archive.org/web/20210813030656/https://clinicaltrials.gov/ct2/show/NCT04078295 (accessed on 13 August 2021).
- Available online: https://web.archive.org/web/20210813031053/https://ascopubs.or (accessed on 13 August 2021).
- Chien, Y.C.; Chou, Y.H.; Wang, W.H.; Chen, J.C.; Chang, W.S.; Tsai, C.W.; Bau, D.T.; Hwang, J.J. Therapeutic Efficacy Evaluation of Pegylated Liposome Encapsulated with Vinorelbine Plus (111) in Repeated Treatments in Human Colorectal Carcinoma with Multimodalities of Molecular Imaging. Cancer Genom. Proteom. 2020, 17, 61–76. [Google Scholar] [CrossRef]
- Greil, R.; Greil-Ressler, S.; Weiss, L.; Schönlieb, C.; Magnes, T.; Radl, B.; Bolger, G.T.; Vcelar, B.; Sordillo, P.P. A phase 1 dose-escalation study on the safety, tolerability and activity of liposomal curcumin (Lipocurc(™)) in patients with locally advanced or metastatic cancer. Cancer Chemother. Pharmacol. 2018, 82, 695–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bolger, G.T.; Licollari, A.; Tan, A.; Greil, R.; Vcelar, B.; Majeed, M.; Helson, L. Distribution and Metabolism of Lipocurc™ (Liposomal Curcumin) in Dog and Human Blood Cells: Species Selectivity and Pharmacokinetic Relevance. Anticancer Res. 2017, 37, 3483. [Google Scholar] [PubMed] [Green Version]
- Available online: https://web.archive.org/web/20210813032817/https://apnews.com/press-release/pr-businesswire/eb9457dc9500491e918b53fdeeb75494 (accessed on 12 August 2021).
- Graziani, S.R.; Vital, C.G.; Morikawa, A.T.; Van Eyll, B.M.; Fernandes Junior, H.J.; Kalil Filho, R.; Maranhão, R.C. Phase II study of paclitaxel associated with lipid core nanoparticles (LDE) as third-line treatment of patients with epithelial ovarian carcinoma. Med. Oncol. 2017, 34, 151. [Google Scholar] [CrossRef] [PubMed]
- Occhiutto, M.L.; Freitas, F.R.; Lima, P.P.; Maranhão, R.C.; Costa, V.P. Paclitaxel Associated with Lipid Nanoparticles as a New Antiscarring Agent in Experimental Glaucoma Surgery. Investig. Ophthalmol. Vis. Sci. 2016, 57, 971–978. [Google Scholar] [CrossRef] [Green Version]
- Gomes, F.L.T.; Maranhão, R.C.; Tavares, E.R.; Carvalho, P.O.; Higuchi, M.L.; Mattos, F.R.; Pitta, F.G.; Hatab, S.A.; Kalil-Filho, R.; Serrano, C.V., Jr. Regression of Atherosclerotic Plaques of Cholesterol-Fed Rabbits by Combined Chemotherapy with Paclitaxel and Methotrexate Carried in Lipid Core Nanoparticles. J. Cardiovasc. Pharmacol. Ther. 2018, 23, 561–569. [Google Scholar] [CrossRef] [PubMed]
- Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007, 2, 751–760. [Google Scholar] [CrossRef]
- Adams, G.P.; Schier, R.; McCall, A.M.; Simmons, H.H.; Horak, E.M.; Alpaugh, R.K.; Marks, J.D.; Weiner, L.M. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Cancer Res. 2001, 61, 4750–4755. [Google Scholar] [PubMed]
- Tolcher, A.W.; Sugarman, S.; Gelmon, K.A.; Cohen, R.; Saleh, M.; Isaacs, C.; Young, L.; Healey, D.; Onetto, N.; Slichenmyer, W. Randomized phase II study of BR96-doxorubicin conjugate in patients with metastatic breast cancer. J. Clin. Oncol. 1999, 17, 478–484. [Google Scholar] [CrossRef] [PubMed]
- Kamoun, W.S.; Kirpotin, D.B.; Huang, Z.R.; Tipparaju, S.K.; Noble, C.O.; Hayes, M.E.; Luus, L.; Koshkaryev, A.; Kim, J.; Olivier, K.; et al. Antitumour activity and tolerability of an EphA2-targeted nanotherapeutic in multiple mouse models. Nat. Biomed. Eng. 2019, 3, 264–280. [Google Scholar] [CrossRef] [PubMed]
- Matusewicz, L.; Filip-Psurska, B.; Psurski, M.; Tabaczar, S.; Podkalicka, J.; Wietrzyk, J.; Ziółkowski, P.; Czogalla, A.; Sikorski, A.F. EGFR-targeted immunoliposomes as a selective delivery system of simvastatin, with potential use in treatment of triple-negative breast cancers. Int. J. Pharm. 2019, 569, 118605. [Google Scholar] [CrossRef] [PubMed]
- Khayrani, A.C.; Mahmud, H.; Oo, A.K.K.; Zahra, M.H.; Oze, M.; Du, J.; Alam, M.J.; Afify, S.M.; Quora, H.A.A.; Shigehiro, T.; et al. Targeting Ovarian Cancer Cells Overexpressing CD44 with Immunoliposomes Encapsulating Glycosylated Paclitaxel. Int. J. Mol. Sci. 2019, 20, 1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hua, S. Synthesis and in vitro characterization of oxytocin receptor targeted PEGylated immunoliposomes for drug delivery to the uterus. J. Liposome Res. 2019, 29, 357–367. [Google Scholar] [CrossRef] [PubMed]
- Takahara, M.; Kamiya, N. Synthetic Strategies for Artificial Lipidation of Functional Proteins. Chem.-A Eur. J. 2020, 26, 4645–4655. [Google Scholar] [CrossRef] [PubMed]
- Marcos-Contreras, O.A.; Greineder, C.F.; Kiseleva, R.Y.; Parhiz, H.; Walsh, L.R.; Zuluaga-Ramirez, V.; Myerson, J.W.; Hood, E.D.; Villa, C.H.; Tombacz, I.; et al. Selective targeting of nanomedicine to inflamed cerebral vasculature to enhance the blood–brain barrier. Proc. Natl. Acad. Sci. USA 2020, 117, 3405. [Google Scholar] [CrossRef] [PubMed]
- Orleth, A.; Mamot, C.; Rochlitz, C.; Ritschard, R.; Alitalo, K.; Christofori, G.; Wicki, A. Simultaneous targeting of VEGF-receptors 2 and 3 with immunoliposomes enhances therapeutic efficacy. J. Drug Target. 2016, 24, 80–89. [Google Scholar] [CrossRef] [PubMed]
- Loureiro, J.A.; Gomes, B.; Fricker, G.; Cardoso, I.; Ribeiro, C.A.; Gaiteiro, C.; Coelho, M.A.; Pereira Mdo, C.; Rocha, S. Dual ligand immunoliposomes for drug delivery to the brain. Colloids Surf. B Biointerfaces 2015, 134, 213–219. [Google Scholar] [CrossRef] [PubMed]
- Paul, J.W.; Hua, S.; Ilicic, M.; Tolosa, J.M.; Butler, T.; Robertson, S.; Smith, R. Drug delivery to the human and mouse uterus using immunoliposomes targeted to the oxytocin receptor. Am. J. Obstet. Gynecol. 2017, 216, e1–e283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moles, E.; Urbán, P.; Jiménez-Díaz, M.B.; Viera-Morilla, S.; Angulo-Barturen, I.; Busquets, M.A.; Fernàndez-Busquets, X. Immunoliposome-mediated drug delivery to Plasmodium-infected and non-infected red blood cells as a dual therapeutic/prophylactic antimalarial strategy. J. Control. Release 2015, 210, 217–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moles, E.; Galiano, S.; Gomes, A.; Quiliano, M.; Teixeira, C.; Aldana, I.; Gomes, P.; Fernàndez-Busquets, X. ImmunoPEGliposomes for the targeted delivery of novel lipophilic drugs to red blood cells in a falciparum malaria murine model. Biomaterials 2017, 145, 178–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramana, L.N.; Sharma, S.; Sethuraman, S.; Ranga, U.; Krishnan, U.M. Stealth anti-CD4 conjugated immunoliposomes with dual antiretroviral drugs--modern Trojan horses to combat HIV. Eur. J. Pharm. Biopharm. 2015, 89, 300–311. [Google Scholar] [CrossRef] [PubMed]
- Ding, T.; Guan, J.; Wang, M.; Long, Q.; Liu, X.; Qian, J.; Wei, X.; Lu, W.; Zhan, C. Natural IgM dominates in vivo performance of liposomes. J. Control. Release 2020, 319, 371–381. [Google Scholar] [CrossRef]
- Rabenhold, M.; Steiniger, F.; Fahr, A.; Kontermann, R.E.; Rüger, R. Bispecific single-chain diabody-immunoliposomes targeting endoglin (CD105) and fibroblast activation protein (FAP) simultaneously. J. Control. Release 2015, 201, 56–67. [Google Scholar] [CrossRef] [PubMed]
- Peng, J.; Chen, J.; Xie, F.; Bao, W.; Xu, H.; Wang, H.; Xu, Y.; Du, Z. Herceptin-conjugated paclitaxel loaded PCL-PEG worm-like nanocrystal micelles for the combinatorial treatment of HER2-positive breast cancer. Biomaterials 2019, 222, 119420. [Google Scholar] [CrossRef]
- Kraft, J.C.; Freeling, J.P.; Wang, Z.; Ho, R.J.Y. Emerging research and clinical development trends of liposome and lipid nanoparticle drug delivery systems. J. Pharm. Sci. 2014, 103, 29–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Available online: https://web.archive.org/web/20210728202637/https://www.sciencenews.org/article/coronavirus-covid-deadly-black-fungus-infection-india (accessed on 28 July 2021).
- Immordino, M.L.; Dosio, F.; Cattel, L. Stealth liposomes: Review of the basic science, rationale, and clinical applications, existing and potential. Int. J. Nanomed. 2006, 1, 297–315. [Google Scholar]
- Scherphof, G.L.; Dijkstra, J.; Spanjer, H.H.; Derksen, J.T.; Roerdink, F.H. Uptake and intracellular processing of targeted and nontargeted liposomes by rat Kupffer cells in vivo and in vitro. Ann. N. Y. Acad. Sci. 1985, 446, 368–384. [Google Scholar] [CrossRef]
- Cullis, P.R.; Chonn, A.; Semple, S.C. Interactions of liposomes and lipid-based carrier systems with blood proteins: Relation to clearance behaviour in vivo. Adv. Drug Deliv. Rev. 1998, 32, 3–17. [Google Scholar]
- Senior, J.H.; Trimble, K.R.; Maskiewicz, R. Interaction of positively-charged liposomes with blood: Implications for their application in vivo. Biochim. Biophys. Acta 1991, 1070, 173–179. [Google Scholar] [CrossRef]
- Oku, N.; Tokudome, Y.; Namba, Y.; Saito, N.; Endo, M.; Hasegawa, Y.; Kawai, M.; Tsukada, H.; Okada, S. Effect of serum protein binding on real-time trafficking of liposomes with different charges analyzed by positron emission tomography. Biochim. Biophys. Acta 1996, 1280, 149–154. [Google Scholar] [CrossRef] [Green Version]
- Philippot, J.R.; Schube, F. Liposomes as Tools in Basic Research and Industry, 1st ed.; CRC Press: Boca Raton, FL, USA, 1994; p. 181. [Google Scholar]
- Du, H.; Chandaroy, P.; Hui, S.W. Grafted poly-(ethylene glycol) on lipid surfaces inhibits protein adsorption and cell adhesion. Biochim. Biophys. Acta 1997, 1326, 236–248. [Google Scholar] [CrossRef] [Green Version]
- Hong, R.L.; Huang, C.J.; Tseng, Y.L.; Pang, V.F.; Chen, S.T.; Liu, J.J.; Chang, F.H. Direct comparison of liposomal doxorubicin with or without polyethylene glycol coating in C-26 tumor-bearing mice: Is surface coating with polyethylene glycol beneficial? Clin. Cancer Res. 1999, 5, 3645–3652. [Google Scholar]
- Parr, M.J.; Masin, D.; Cullis, P.R.; Bally, M.B. Accumulation of liposomal lipid and encapsulated doxorubicin in murine Lewis lung carcinoma: The lack of beneficial effects by coating liposomes with poly(ethylene-glycol). J. Pharmacol. Exp. Ther. 1997, 280, 1319–1327. [Google Scholar]
- Kale, A.A.; Torchilin, V.P. Design, Synthesis, and Characterization of pH-Sensitive PEG−PE Conjugates for Stimuli-Sensitive Pharmaceutical Nanocarriers: The Effect of Substitutes at the Hydrazone Linkage on the pH Stability of PEG−PE Conjugates. Bioconjugate Chem. 2007, 18, 363–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Y.; Zhao, D.; Wang, G.; Wang, Y.; Cao, L.; Sun, J.; Jiang, Q.; He, Z. Recent progress of hypoxia-modulated multifunctional nanomedicines to enhance photodynamic therapy: Opportunities, challenges, and future development. Acta Pharm. Sin. B 2020, 10, 1382–1396. [Google Scholar] [CrossRef]
- Sharma, A.; Arambula, J.F.; Koo, S.; Kumar, R.; Singh, H.; Sessler, J.L.; Kim, J.S. Hypoxia-targeted drug delivery. Chem. Soc. Rev. 2019, 48, 771–813. [Google Scholar] [CrossRef]
- Andresen, T.L.; Thompson, D.H.; Kaasgaard, T. Enzyme-triggered nanomedicine: Drug release strategies in cancer therapy (Invited Review). Mol. Membr. Biol. 2010, 27, 353–363. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Zou, Q.; Xing, R.; Govindaraju, T.; Fakhrullin, R.; Yan, X. Peptide-modulated self-assembly as a versatile strategy for tumor supramolecular nanotheranostics. Theranostics 2019, 9, 3249–3261. [Google Scholar] [CrossRef] [PubMed]
- Horsman, M.R.; Vaupel, P. Pathophysiological Basis for the Formation of the Tumor Microenvironment. Front. Oncol. 2016, 6, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalafatovic, D.; Nobis, M.; Son, J.; Anderson, K.I.; Ulijn, R.V. MMP-9 triggered self-assembly of doxorubicin nanofiber depots halts tumor growth. Biomaterials 2016, 98, 192–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, N.; Zhao, F.; Zou, Q.; Li, Y.; Ma, G.; Yan, X. Multitriggered Tumor-Responsive Drug Delivery Vehicles Based on Protein and Polypeptide Coassembly for Enhanced Photodynamic Tumor Ablation. Small 2016, 12, 5936–5943. [Google Scholar] [CrossRef]
- Canavese, G.; Ancona, A.; Racca, L.; Canta, M.; Dumontel, B.; Barbaresco, F.; Limongi, T.; Cauda, V. Nanoparticle-assisted ultrasound: A special focus on sonodynamic therapy against cancer. Chem. Eng. J. 2018, 340, 155–172. [Google Scholar] [CrossRef]
- Sirsi, S.R.; Borden, M.A. State-of-the-art materials for ultrasound-triggered drug delivery. Adv. Drug Deliv. Rev. 2014, 72, 3–14. [Google Scholar] [CrossRef] [Green Version]
- VanOsdol, J.; Ektate, K.; Ramasamy, S.; Maples, D.; Collins, W.; Malayer, J.; Ranjan, A. Sequential HIFU heating and nanobubble encapsulation provide efficient drug penetration from stealth and temperature sensitive liposomes in colon cancer. J. Control. Release 2017, 247, 55–63. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.L.; MacDonald, R.C. Acoustically active liposomes for drug encapsulation and ultrasound-triggered release. Biochim. Biophys. Acta 2004, 1665, 134–141. [Google Scholar] [CrossRef] [Green Version]
- Rapoport, N. Combined cancer therapy by micellar-encapsulated drug and ultrasound. Int. J. Pharm. 2004, 277, 155–162. [Google Scholar] [CrossRef] [PubMed]
- Leung, S.J.; Romanowski, M. Light-Activated Content Release from Liposomes. Theranostics 2012, 2, 1020–1036. [Google Scholar] [CrossRef]
- Pidgeon, C.; Hunt, C.A. Light sensitive liposomes. Photochem. Photobiol. 1983, 37, 491–494. [Google Scholar] [CrossRef]
- Ohya, Y.; Okuyama, Y.; Fukunaga, A.; Ouchi, T. Photo-sensitive lipid membrane perturbation by a single chain lipid having terminal spiropyran group. Supramol. Chem. 1998, 5, 21–29. [Google Scholar] [CrossRef]
- Anderson, V.C.; Thompson, D.H. Triggered release of hydrophilic agents from plasmalogen liposomes using visible light or acid. Biochim. Biophys. Acta 1992, 1109, 33–42. [Google Scholar] [CrossRef]
- Thompson, D.H.; Gerasimov, O.V.; Wheeler, J.J.; Rui, Y.; Anderson, V.C. Triggerable plasmalogen liposomes: Improvement of system efficiency. Biochim. Biophys. Acta 1996, 1279, 25–34. [Google Scholar] [CrossRef] [Green Version]
- Wan, Y.; Angleson, J.K.; Kutateladze, A.G. Liposomes from Novel Photolabile Phospholipids: Light-Induced Unloading of Small Molecules as Monitored by PFG NMR. J. Am. Chem. Soc. 2002, 124, 5610–5611. [Google Scholar] [CrossRef]
- Li, Z.; Wan, Y.; Kutateladze, A.G. Dithiane-based photolabile amphiphiles: Toward photolabile liposomes. Langmuir 2003, 19, 6381–6391. [Google Scholar] [CrossRef]
- Zhang, Z.; Smith, B.D. Synthesis and Characterization of NVOC-DOPE, a Caged Photoactivatable Derivative of Dioleoylphosphatidylethanolamine. Bioconjugate Chem. 1999, 10, 1150–1152. [Google Scholar] [CrossRef] [PubMed]
- Bayer, A.M.; Alam, S.; Mattern-Schain, S.I.; Best, M.D. Triggered liposomal release through a synthetic phosphatidylcholine analogue bearing a photocleavable moiety embedded within the sn-2 acyl chain. Chemistry 2014, 20, 3350–3357. [Google Scholar] [CrossRef]
- O’Brien, D.F.; Armitage, B.; Benedicto, A.; Bennett, D.E.; Lamparski, H.G.; Lee, Y.; Srisiri, W.; Sisson, T.M. Polymerization of Preformed Self-Organized Assemblies. Acc. Chem. Res. 1998, 31, 861–868. [Google Scholar] [CrossRef]
- Yavlovich, A.; Singh, A.; Blumenthal, R.; Puri, A. A novel class of photo-triggerable liposomes containing DPPC:DC8,9PC as vehicles for delivery of doxorubcin to cells. Biochim. Biophys. Acta Biomembr. 2011, 1808, 117–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gerasimov, O.V.; Boomer, J.A.; Qualls, M.M.; Thompson, D.H. Cytosolic drug delivery using pH- and light-sensitive liposomes. Adv. Drug Deliv. Rev. 1999, 38, 317–338. [Google Scholar] [CrossRef]
- Fuse, T.; Tagami, T.; Tane, M.; Ozeki, T. Effective light-triggered contents release from helper lipid-incorporated liposomes co-encapsulating gemcitabine and a water-soluble photosensitizer. Int. J. Pharm. 2018, 540, 50–56. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Li, W.; Di, H.; Luo, L.; Zhu, C.; Yang, J.; Yin, X.; Yin, H.; Gao, J.; Du, Y.; et al. A photosensitive liposome with NIR light triggered doxorubicin release as a combined photodynamic-chemo therapy system. J. Control. Release 2018, 277, 114–125. [Google Scholar] [CrossRef]
- Yatvin, M.B.; Weinstein, J.N.; Dennis, W.H.; Blumenthal, R. Design of liposomes for enhanced local release of drugs by hyperthermia. Science 1978, 202, 1290–1293. [Google Scholar] [CrossRef]
- Ta, T.; Porter, T.M. Thermosensitive liposomes for localized delivery and triggered release of chemotherapy. J. Control. Release 2013, 169, 112–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nibu, Y.; Inoue, T.; Motoda, I. Effect of headgroup type on the miscibility of homologous phospholipids with different acyl chain lengths in hydrated bilayer. Biophys. Chem. 1995, 56, 273–280. [Google Scholar] [CrossRef]
- Meshorer, A.; Prionas, S.D.; Fajardo, L.F.; Meyer, J.L.; Hahn, G.M.; Martinez, A.A. The effects of hyperthermia on normal mesenchymal tissues. Application of a histologic grading system. Arch. Pathol. Lab. Med. 1983, 107, 328–334. [Google Scholar] [PubMed]
- Anyarambhatla, G.R.; Needham, D. Enhancement of the Phase Transition Permeability of DPPC Liposomes by Incorporation of MPPC: A New Temperature-Sensitive Liposome for use with Mild Hyperthermia. J. Liposome Res. 1999, 9, 491–506. [Google Scholar] [CrossRef]
- Banno, B.; Ickenstein, L.M.; Chiu, G.N.C.; Bally, M.B.; Thewalt, J.; Brief, E.; Wasan, E.K. The functional roles of poly(ethylene glycol)-lipid and lysolipid in the drug retention and release from lysolipid-containing thermosensitive liposomes in vitro and in vivo. J. Pharm. Sci. 2010, 99, 2295–2308. [Google Scholar] [CrossRef] [PubMed]
- Sandström, M.C.; Ickenstein, L.M.; Mayer, L.D.; Edwards, K. Effects of lipid segregation and lysolipid dissociation on drug release from thermosensitive liposomes. J. Control. Release 2005, 107, 131–142. [Google Scholar] [CrossRef] [PubMed]
- Available online: https://web.archive.org/web/20210728223201/https://www.globenewswire.com/en/news-release/2020/04/15/2016425/0/en/Celsion-Reports-that-Sufficient-Events-Have-Been-Reached-for-the-Second-Interim-Analysis-of-the-Phase-III-OPTIMA-Study-of-ThermoDox-in-Primary-Liver-Cancer.html (accessed on 28 July 2021).
- Swenson, C.E.; Haemmerich, D.; Maul, D.H.; Knox, B.; Ehrhart, N.; Reed, R.A. Increased Duration of Heating Boosts Local Drug Deposition during Radiofrequency Ablation in Combination with Thermally Sensitive Liposomes (ThermoDox) in a Porcine Model. PLoS ONE 2015, 10, e0139752. [Google Scholar] [CrossRef]
- Amstad, E.; Kohlbrecher, J.; Müller, E.; Schweizer, T.; Textor, M.; Reimhult, E. Triggered Release from Liposomes through Magnetic Actuation of Iron Oxide Nanoparticle Containing Membranes. Nano Lett. 2011, 11, 1664–1670. [Google Scholar] [CrossRef]
- Babincová, M.; Cicmanec, P.; Altanerová, V.; Altaner, C.; Babinec, P. AC-magnetic field controlled drug release from magnetoliposomes: Design of a method for site-specific chemotherapy. Bioelectrochemistry 2002, 55, 17–19. [Google Scholar] [CrossRef]
- Nobuto, H.; Sugita, T.; Kubo, T.; Shimose, S.; Yasunaga, Y.; Murakami, T.; Ochi, M. Evaluation of systemic chemotherapy with magnetic liposomal doxorubicin and a dipole external electromagnet. Int. J. Cancer 2004, 109, 627–635. [Google Scholar] [CrossRef]
- Kubo, T.; Sugita, T.; Shimose, S.; Nitta, Y.; Ikuta, Y.; Murakami, T. Targeted systemic chemotherapy using magnetic liposomes with incorporated adriamycin for osteosarcoma in hamsters. Int. J. Oncol. 2001, 18, 121–125. [Google Scholar] [CrossRef] [PubMed]
- Babincová, M.; Altanerová, V.; Lampert, M.; Altaner, C.; Machová, E.; Srámka, M.; Babinec, P. Site-specific in vivo targeting of magnetoliposomes using externally applied magnetic field. Z. Naturforsch. C J. Biosci. 2000, 55, 278–281. [Google Scholar] [CrossRef]
- Matsunaga, T.; Higashi, Y.; Tsujimura, N. Drug delivery by magnetoliposomes containing bacterial magnetic particles. Cell Eng. 1997, 2, 7–11. [Google Scholar]
- Jain, S.; Mishra, V.; Singh, P.; Dubey, P.K.; Saraf, D.K.; Vyas, S.P. RGD-anchored magnetic liposomes for monocytes/neutrophils-mediated brain targeting. Int. J. Pharm. 2003, 261, 43–55. [Google Scholar] [CrossRef]
- Smith, J.A.; Costales, A.B.; Jaffari, M.; Urbauer, D.L.; Frumovitz, M.; Kutac, C.K.; Tran, H.; Coleman, R.L. Is it equivalent? Evaluation of the clinical activity of single agent Lipodox® compared to single agent Doxil® in ovarian cancer treatment. J. Oncol. Pharm. Pract. 2016, 22, 599–604. [Google Scholar] [CrossRef]
- Bekersky, I.; Fielding, R.M.; Dressler, D.E.; Lee, J.W.; Buell, D.N.; Walsh, T.J. Pharmacokinetics, excretion, and mass balance of liposomal amphotericin B (AmBisome) and amphotericin B deoxycholate in humans. Antimicrob. Agents Chemother. 2002, 46, 828–833. [Google Scholar] [CrossRef] [Green Version]
- Charrois, G.J.R.; Allen, T.M. Drug release rate influences the pharmacokinetics, biodistribution, therapeutic activity, and toxicity of pegylated liposomal doxorubicin formulations in murine breast cancer. Biochim. Biophys. Acta 2004, 1663, 167–177. [Google Scholar] [CrossRef]
- Boswell, G.W.; Buell, D.; Bekersky, I. AmBisome (liposomal amphotericin B): A comparative review. J. Clin. Pharmacol. 1998, 38, 583–592. [Google Scholar] [CrossRef]
- Walsh, T.J.; Yeldandi, V.; McEvoy, M.; Gonzalez, C.; Chanock, S.; Freifeld, A.; Seibel, N.I.; Whitcomb, P.O.; Jarosinski, P.; Boswell, G.; et al. Safety, Tolerance, and Pharmacokinetics of a Small Unilamellar Liposomal Formulation of Amphotericin B (AmBisome) in Neutropenic Patients. Antimicrob. Agents Chemother. 1998, 42, 2391–2398. [Google Scholar] [CrossRef] [Green Version]
- Paolo, D.D.; Ambrogio, C.; Pastorino, F.; Brignole, C.; Martinengo, C.; Carosio, R.; Loi, M.; Pagnan, G.; Emionite, L.; Cilli, M.; et al. Selective Therapeutic Targeting of the Anaplastic Lymphoma Kinase with Liposomal siRNA Induces Apoptosis and Inhibits Angiogenesis in Neuroblastoma. Mol. Ther. 2011, 19, 2201–2212. [Google Scholar] [CrossRef]
- Cui, J.; Li, C.; Guo, W.; Li, Y.; Wang, C.; Zhang, L.; Zhang, L.; Hao, Y.; Wang, Y. Direct comparison of two pegylated liposomal doxorubicin formulations: Is AUC predictive for toxicity and efficacy? J. Control. Release 2007, 118, 204–215. [Google Scholar] [CrossRef]
- Cheung, B.C.L.; Sun, T.H.T.; Leenhouts, J.M.; Cullis, P.R. Loading of doxorubicin into liposomes by forming Mn2+-drug complexes. Biochim. Biophys. Acta Biomembr. 1998, 1414, 205–216. [Google Scholar] [CrossRef] [Green Version]
- Szebeni, J.; Storm, G. Complement activation as a bioequivalence issue relevant to the development of generic liposomes and other nanoparticulate drugs. Biochem. Biphys. Res. Commun. 2015, 468, 490–497. [Google Scholar] [CrossRef]
- Szebeni, J.; Fishbane, S.; Hedenus, M.; Howaldt, S.; Locatelli, F.; Patni, S.; Rampton, D.; Weiss, G.; Folkersen, J. Hypersensitivity to intravenous iron: Classification, terminology, mechanisms and management. Br. J. Pharmacol. 2015, 172, 5025–5036. [Google Scholar] [CrossRef] [Green Version]
- Szebeni, J.; Muggia, F.; Barenholz, Y. Case Study: Complement Activation Related Hypersensitivity Reactions to PEGylated Liposomal Doxorubicin—Experimental and Clinical Evidence, Mechanisms and Approaches to Inhibition. In Handbook of Immunological Properties of Engineered Nanomaterials, 2nd ed.; Dobrovolskaia, M.A., McNeil, S.E., Eds.; World Scientific Publishing Co Pte Ltd.: Singapore, 2016; Volume 2, pp. 331–361. [Google Scholar]
- Kelly, C.; Lawlor, C.; Burke, C.; Barlow, J.W.; Ramsey, J.M.; Jefferies, C.; Cryan, S. High-throughput methods for screening liposome-macrophage cell interaction. J. Liposome Res. 2015, 25, 211–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wientjes, M.G.; Yeung, B.Z.; Lu, Z.; Wientjes, M.G.; Au, J.L.S. Predicting diffusive transport of cationic liposomes in 3-dimensional tumor spheroids. J. Control. Release 2014, 192, 10–18. [Google Scholar] [CrossRef] [Green Version]
Active Ingredient/s | Trade/Brand Name | Liposome Composition | Active/Passive Targeting | Route of Administration | Indication | Ref. |
---|---|---|---|---|---|---|
Hwtp53 DNA | SGT-53 | DOTAP/DOPE | Active (Anti-Transferrin scFv) | IV, in vivo, clinical | Solid tumors | [13,14,15] |
Docetaxel prodrug | MM-310 | Egg derived sphingomyelin/ CH | Active (Anti-Ephrin receptor A2) | IV, in vivo, clinical | Solid tumors | [16,17,18,19] |
DOX | C225-ILs-dox | DSPC/CH/mPEG-DSPE | Active (Anti-EGFR Fab fragment from mAb C225 (cetuximab)) | IV, in vivo, clinical | Glioblastoma | [15,19,20,21] |
DOX | MM-302 | HSPC/CH/DSPE-PEG | Active (Anti-HER2 antibody) | IV, in vivo, clinical | Breast cancer | [22,23] |
Melanoma antigens + interferon-gamma | Lipovaxin-MM | POPC/Ni-3NTA-DTDA | Active (Single domain antibody (dAb) fragment (VH)) | IV, in vivo, clinical | Malignant melanoma | [15,24] |
RB94 plasmid DNA | SGT-94 | DOTAP/DOPE | Active (Anti-Transferrin Antibody fragment (scFv)) | IV, in vivo, clinical | Solid tumor | [15,25,26,27] |
DOX | 2B3-101 | HSPC/CH/DSPE-PEG | Active (Glutathione ligand) | IV, in vivo, clinical | Active brain metastasis, meningeal carcinomatosis | [18,28,29] |
Tetrandrine + vincristine | - | EPC/CH/DSPE-PEG 2000 | Active (Transferrin ligand) | IV, in vivo in mice | Brain glioma | [19,30] |
Bleomycin | - | DOPE/CH | Active (Folic acid ligand) | In vitro | Cervical and breast cancer cell lines | [19,31] |
DOX | - | DOPE/DOPC/Lecithin | Active (Glycoprotein ligand) | IV, in vivo in mice | Mouse melanoma cells | [32] |
ATRA | - | DPPC/CH/DSPE-mPEG2000 | Passive | In vitro | Human thyroid carcinoma cell lines | [33] |
ATRA | - | DOTAP/CH | Passive | In vivo in mice, IV | Lung cancer | [34] |
Daunorubicin + Cytarabine | VYXEOS | DSPG/DSPC/CH | Passive | IV, in vivo, FDA approved | Secondary acute myeloid leukemia (sAML) | [15,35,36,37] |
Paclitaxel | LEP-ETU | DOPC/CH/cardiolipin | Passive | IV, in vivo, FDA approved | Ovarian cancer | [38,39] |
Vincristine | - | Sphingomyelin/CH | Passive | IV, in vivo, clinical | Philadelphia chromosome-negative (Ph-) acute lymphoblastic leukemia (ALL) | [40,41,42] |
Verteporfin | Visudyne | DMPC/EPG | Passive | IV, in vivo, clinical | EGFR-mutated glioblastoma | [43,44,45] |
DOX | ThermoDox | DPPC/MSPC/PEG 2000-DSPE | Passive | IV, in vivo, clinical | Hepatocellular carcinoma (HCC) | [46,47] |
Paclitaxel | EndoTAG-1 | DOTAP/DOPC | Passive | IV, in vivo, clinical | Pancreatic cancer | [38,48] |
miR-34a | - | DOTAP/CH | Passive | IV, in vivo, clinical | Advanced solid tumors | [40,49,50,51] |
Irinotecan | ONIVYDE | DSPC/DSPE/CH/mPEG-2000 | Passive | IV, in vivo, FDA approved | Metastatic adenocarcinoma of the pancreas | [52,53] |
Mitomycin-C prodrug | Promitil | HSPC/CH/DSPE-PEG | Passive | IV, in vivo, clinical | Solid tumors | [54,55,56] |
TUSC2/FUS1 | REQORSA | DOTAP/CH | Passive | IV, in vivo, clinical | Non-Small cell lung cancer | [57,58] |
Eribulin mesylate | E7389-LF | HSPC/CH/PEG 2000-DSPE | Passive | IV, in vivo, clinical | Solid tumors | [15,59,60] |
Navelbine | - | DSPC/CH/PEG -DSPE | Passive | In vivo in mice | Colorectal cancer cells | [61] |
Curcumin | Lipocurc | DMPG/DMPC | Passive | IV, in vivo, clinical | Metastatic tumors | [62,63,64] |
Paclitaxel | PTX–LDE | Cholesteryl oleate/Egg-PC/Miglyol 812/CH | Passive | IV, in vivo, clinical | Ovarian carcinoma | [65,66,67] |
PKN3 siRNA | Atu027 | AtuFECT01/DPhyPE/DSPE-PEG-2000 | Passive | IV, in vivo, clinical | Pancreatic cancer | [25] |
Active and Passive Targeting Challenges | |
---|---|
1. | Scale up liposome preparation to reproducibly achieve target product profile including in vitro drug release rate, particle size distribution, lamellarity, stability, drug encapsulation efficiency, etc. |
2. | Separation of raw lipids in a mixture of lipids and ability to analyze them |
3. | Determination of complete stability and toxicity profile of novel lipids involved in formulations |
4. | Stability of liposomes in solution |
5. | Determination of biodistribution of liposomes appropriate PK/PD models to predict parameters in humans |
6. | Immunogenic reactions such as CARPA upon IV administration of liposomes have resulted in additional layer of challenge |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gyanani, V.; Haley, J.C.; Goswami, R. Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems. Pharmaceuticals 2021, 14, 835. https://doi.org/10.3390/ph14090835
Gyanani V, Haley JC, Goswami R. Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems. Pharmaceuticals. 2021; 14(9):835. https://doi.org/10.3390/ph14090835
Chicago/Turabian StyleGyanani, Vijay, Jeffrey C. Haley, and Roshan Goswami. 2021. "Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems" Pharmaceuticals 14, no. 9: 835. https://doi.org/10.3390/ph14090835
APA StyleGyanani, V., Haley, J. C., & Goswami, R. (2021). Challenges of Current Anticancer Treatment Approaches with Focus on Liposomal Drug Delivery Systems. Pharmaceuticals, 14(9), 835. https://doi.org/10.3390/ph14090835